Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Br J Clin Pharmacol ; 88(6): 2525-2538, 2022 06.
Article in English | MEDLINE | ID: mdl-34022071

ABSTRACT

RNA interference (RNAi) is a natural biological pathway that inhibits gene expression by targeted degradation or translational inhibition of cytoplasmic mRNA by the RNA induced silencing complex. RNAi has long been exploited in laboratory research to study the biological consequences of the reduced expression of a gene of interest. More recently RNAi has been demonstrated as a therapeutic avenue for rare metabolic diseases. This review presents an overview of the cellular RNAi machinery as well as therapeutic RNAi design and delivery. As a clinical example we present primary hyperoxaluria, an ultrarare inherited disease of increased hepatic oxalate production which leads to recurrent calcium oxalate kidney stones. In the most common form of the disease (Type 1), end-stage kidney disease occurs in childhood or young adulthood, often necessitating combined kidney and liver transplantation. In this context we discuss nedosiran (Dicerna Pharmaceuticals, Inc.) and lumasiran (Alnylam Pharmaceuticals), which are both novel RNAi therapies for primary hyperoxaluria that selectively reduce hepatic expression of lactate dehydrogenase and glycolate oxidase respectively, reducing hepatic oxalate production and urinary oxalate levels. Finally, we consider future optimizations advances in RNAi therapies.


Subject(s)
Hyperoxaluria, Primary , RNA Interference , Adult , Female , Humans , Hyperoxaluria, Primary/genetics , Hyperoxaluria, Primary/metabolism , Hyperoxaluria, Primary/therapy , Male , Oxalates/metabolism , Oxalates/urine , RNA, Small Interfering , Young Adult
2.
Mol Ther ; 26(8): 1983-1995, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29914758

ABSTRACT

Primary hyperoxalurias (PHs) are autosomal recessive disorders caused by the overproduction of oxalate leading to calcium oxalate precipitation in the kidney and eventually to end-stage renal disease. One promising strategy to treat PHs is to reduce the hepatic production of oxalate through substrate reduction therapy by inhibiting liver-specific glycolate oxidase (GO), which controls the conversion of glycolate to glyoxylate, the proposed main precursor to oxalate. Alternatively, diminishing the amount of hepatic lactate dehydrogenase (LDH) expression, the proposed key enzyme responsible for converting glyoxylate to oxalate, should directly prevent the accumulation of oxalate in PH patients. Using RNAi, we provide the first in vivo evidence in mammals to support LDH as the key enzyme responsible for converting glyoxylate to oxalate. In addition, we demonstrate that reduction of hepatic LDH achieves efficient oxalate reduction and prevents calcium oxalate crystal deposition in genetically engineered mouse models of PH types 1 (PH1) and 2 (PH2), as well as in chemically induced PH mouse models. Repression of hepatic LDH in mice did not cause any acute elevation of circulating liver enzymes, lactate acidosis, or exertional myopathy, suggesting further evaluation of liver-specific inhibition of LDH as a potential approach for treating PH1 and PH2 is warranted.


Subject(s)
Hyperoxaluria, Primary/therapy , L-Lactate Dehydrogenase/antagonists & inhibitors , Oxalates/metabolism , RNA Interference/physiology , Animals , Disease Models, Animal , Gene Silencing , Humans , Hyperoxaluria, Primary/genetics , Hyperoxaluria, Primary/metabolism , L-Lactate Dehydrogenase/genetics , Liver/enzymology , Mice
3.
Mol Ther ; 26(7): 1771-1782, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29784585

ABSTRACT

Glycogen storage diseases (GSDs) of the liver are devastating disorders presenting with fasting hypoglycemia as well as hepatic glycogen and lipid accumulation, which could lead to long-term liver damage. Diet control is frequently utilized to manage the potentially dangerous hypoglycemia, but there is currently no effective pharmacological treatment for preventing hepatomegaly and concurrent liver metabolic abnormalities, which could lead to fibrosis, cirrhosis, and hepatocellular adenoma or carcinoma. In this study, we demonstrate that inhibition of glycogen synthesis using an RNAi approach to silence hepatic Gys2 expression effectively prevents glycogen synthesis, glycogen accumulation, hepatomegaly, fibrosis, and nodule development in a mouse model of GSD III. Mechanistically, reduction of accumulated abnormally structured glycogen prevents proliferation of hepatocytes and activation of myofibroblasts as well as infiltration of mononuclear cells. Additionally, we show that silencing Gys2 expression reduces hepatic steatosis in a mouse model of GSD type Ia, where we hypothesize that the reduction of glycogen also reduces the production of excess glucose-6-phosphate and its subsequent diversion to lipid synthesis. Our results support therapeutic silencing of GYS2 expression to prevent glycogen and lipid accumulation, which mediate initial signals that subsequently trigger cascades of long-term liver injury in GSDs.


Subject(s)
Glycogen Storage Disease Type III/genetics , Glycogen Synthase/genetics , Glycogen/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver/pathology , RNA Interference/physiology , Animals , Disease Models, Animal , Female , Fibroblasts/pathology , Glucose-6-Phosphate/genetics , Glycogen Storage Disease Type III/pathology , Hepatocytes/pathology , Hepatomegaly/genetics , Male , Mice , Mice, Inbred C57BL
4.
Mol Cancer Ther ; 17(2): 544-553, 2018 02.
Article in English | MEDLINE | ID: mdl-29282298

ABSTRACT

Colorectal carcinomas harbor well-defined genetic abnormalities, including aberrant activation of Wnt/ß-catenin and MAPK pathways, often simultaneously. Although the MAPK pathway can be targeted using potent small-molecule drugs, including BRAF and MEK inhibitors, ß-catenin inhibition has been historically challenging. RNAi approaches have advanced to the stage of clinical viability and are especially well suited for transcriptional modulators, such as ß-catenin. In this study, we report therapeutic effects of combined targeting of these pathways with pharmacologic agents. Using a recently described tumor-selective nanoparticle containing a ß-catenin-targeting RNAi trigger, in combination with the FDA-approved MEK inhibitor (MEKi) trametinib, we demonstrate synergistic tumor growth inhibition in in vivo models of colorectal cancer, melanoma, and hepatocellular carcinoma. At dose levels that were insufficient to significantly impact tumor growth as monotherapies, combination regimens resulted in synergistic efficacy and complete tumor growth inhibition. Importantly, dual MEKi/RNAi therapy dramatically improved survival of mice bearing colorectal cancer liver metastases. In addition, pharmacologic silencing of ß-catenin mRNA was effective against tumors that are inherently resistant or that acquire drug-induced resistance to trametinib. These results provide a strong rationale for clinical evaluation of this dual-targeting approach for cancers harboring Wnt/ß-catenin and MAPK pathway mutations. Mol Cancer Ther; 17(2); 544-53. ©2017 AACR.


Subject(s)
Colorectal Neoplasms/therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , beta Catenin/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Synergism , Gene Silencing , Heterografts , Humans , Liver Neoplasms, Experimental/secondary , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Nanoparticles/administration & dosage , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
5.
Mol Cancer Ther ; 15(9): 2143-54, 2016 09.
Article in English | MEDLINE | ID: mdl-27390343

ABSTRACT

The Wnt/ß-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/ß-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding ß-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143-54. ©2016 AACR.


Subject(s)
Neoplasms/genetics , RNA Interference , RNA, Small Interfering/genetics , beta Catenin/genetics , Animals , Apolipoproteins E/chemistry , Apolipoproteins E/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lipids/chemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Melanoma, Experimental , Mice , Nanoparticles/chemistry , Neoplasm Metastasis , Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Structure-Activity Relationship , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism , ras Proteins/genetics , ras Proteins/metabolism
6.
Mol Ther ; 24(4): 770-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26758691

ABSTRACT

Primary hyperoxaluria type 1 (PH1) is an autosomal recessive, metabolic disorder caused by mutations of alanine-glyoxylate aminotransferase (AGT), a key hepatic enzyme in the detoxification of glyoxylate arising from multiple normal metabolic pathways to glycine. Accumulation of glyoxylate, a precursor of oxalate, leads to the overproduction of oxalate in the liver, which accumulates to high levels in kidneys and urine. Crystalization of calcium oxalate (CaOx) in the kidney ultimately results in renal failure. Currently, the only treatment effective in reduction of oxalate production in patients who do not respond to high-dose vitamin B6 therapy is a combined liver/kidney transplant. We explored an alternative approach to prevent glyoxylate production using Dicer-substrate small interfering RNAs (DsiRNAs) targeting hydroxyacid oxidase 1 (HAO1) mRNA which encodes glycolate oxidase (GO), to reduce the hepatic conversion of glycolate to glyoxylate. This approach efficiently reduces GO mRNA and protein in the livers of mice and nonhuman primates. Reduction of hepatic GO leads to normalization of urine oxalate levels and reduces CaOx deposition in a preclinical mouse model of PH1. Our results support the use of DsiRNA to reduce liver GO levels as a potential therapeutic approach to treat PH1.


Subject(s)
Alcohol Oxidoreductases/genetics , Calcium Oxalate/metabolism , Hyperoxaluria, Primary/therapy , RNA, Small Interfering/administration & dosage , Animals , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Glyoxylates/urine , Humans , Hyperoxaluria, Primary/enzymology , Hyperoxaluria, Primary/urine , Liver/metabolism , Mice , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Ribonuclease III/metabolism
7.
Mol Cancer Ther ; 12(6): 925-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536719

ABSTRACT

CUDC-101 is a novel, small-molecule, anticancer agent targeting histone deacetylase (HDAC), EGF receptor (EGFR), and HER2. It is currently in phase I clinical development in patients with solid tumors. Previously, we reported that CUDC-101 has potent antiproliferative and proapoptotic activity in cultured tumor cells and in vivo xenograft models. We now show that cancer cells that have acquired resistance to single-target EGFR inhibitors through upregulation of AXL or loss of E-cadherin remain sensitive to CUDC-101, which inhibits MET- and AXL-mediated signaling, restores E-cadherin expression, and reduces cell migration. CUDC-101 also efficiently inhibited the proliferation of MET-overexpressing non-small cell lung cancer and gastric cancer cell lines and inhibited the migration and invasion of invasive tumor cells. Taken together, these results suggest that coupling HDAC and HER2 inhibitory activities to an EGFR inhibitor may potentially be effective in overcoming drug resistance and preventing cancer cell migration.


Subject(s)
ErbB Receptors/metabolism , Histone Deacetylases/metabolism , Hydroxamic Acids/administration & dosage , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/administration & dosage , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
8.
Clin Cancer Res ; 18(15): 4104-13, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22693356

ABSTRACT

PURPOSE: Given that histone deacetylase (HDAC) inhibitors are known to induce multiple epigenetic modifications affecting signaling networks and act synergistically with phosphatidylinositol 3-kinase (PI3K) inhibitors, we developed a strategy to simultaneously inhibit HDACs and PI3K in cancer cells. EXPERIMENTAL DESIGN: We constructed dual-acting inhibitors by incorporating HDAC inhibitory functionality into a PI3K inhibitor pharmacophore. CUDC-907, a development candidate selected from these dual inhibitors, was evaluated in vitro and in vivo to determine its pharmacologic properties, anticancer activity, and mechanism of action. RESULTS: CUDC-907 potently inhibits class I PI3Ks as well as classes I and II HDAC enzymes. Through its integrated HDAC inhibitory activity, CUDC-907 durably inhibits the PI3K-AKT-mTOR pathway and compensatory signaling molecules such as RAF, MEK, MAPK, and STAT-3, as well as upstream receptor tyrosine kinases. CUDC-907 shows greater growth inhibition and proapoptotic activity than single-target PI3K or HDAC inhibitors in both cultured and implanted cancer cells. CONCLUSIONS: CUDC-907 may offer improved therapeutic benefits through simultaneous, sustained disruption of multiple oncogenic signaling networks.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylases/metabolism , Morpholines/pharmacology , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , HCT116 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mice , Mice, Nude , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Sf9 Cells , Tumor Burden/drug effects , Vorinostat , Xenograft Model Antitumor Assays
9.
Cancer Res ; 70(9): 3647-56, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388807

ABSTRACT

Receptor tyrosine kinase inhibitors have recently become important therapeutics for a variety of cancers. However, due to the heterogeneous and dynamic nature of tumors, the effectiveness of these agents is often hindered by poor response rates and acquired drug resistance. To overcome these limitations, we created a novel small molecule, CUDC-101, which simultaneously inhibits histone deacetylase and the receptor kinases epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) in cancer cells. Because of its integrated histone deacetylase inhibition, CUDC-101 synergistically blocked key regulators of EGFR/HER2 signaling pathways, also attenuating multiple compensatory pathways, such as AKT, HER3, and MET, which enable cancer cells to escape the effects of conventional EGFR/HER2 inhibitors. CUDC-101 displayed potent antiproliferative and proapoptotic activities against cultured and implanted tumor cells that are sensitive or resistant to several approved single-targeted drugs. Our results show that CUDC-101 has the potential to dramatically improve the treatment of heterogeneous and drug-resistant tumors that cannot be controlled with single-target agents. Further, they provide a framework to create individual small molecules that simultaneously antagonize multiple biochemically distinct oncogenic targets, suggesting a general paradigm to surpass conventional, single-target cancer therapeutics. Cancer Res; 70(9); 3647-56. (c)2010 AACR.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
J Med Chem ; 53(5): 2000-9, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20143778

ABSTRACT

By incorporating histone deacetylase (HDAC) inhibitory functionality into the pharmacophore of the epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) inhibitors, we synthesized a novel series of compounds with potent, multiacting HDAC, EGFR, and HER2 inhibition and identified 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide 8 (CUDC-101) as a drug candidate, which is now in clinical development. 8 displays potent in vitro inhibitory activity against HDAC, EGFR, and HER2 with an IC(50) of 4.4, 2.4, and 15.7 nM, respectively. In most tumor cell lines tested, 8 exhibits efficient antiproliferative activity with greater potency than vorinostat (SAHA), erlotinib, lapatinib, and combinations of vorinostat/erlotinib and vorinostat/lapatinib. In vivo, 8 promotes tumor regression or inhibition in various cancer xenograft models including nonsmall cell lung cancer (NSCLC), liver, breast, head and neck, colon, and pancreatic cancers. These results suggest that a single compound that simultaneously inhibits HDAC, EGFR, and HER2 may offer greater therapeutic benefits in cancer over single-acting agents through the interference with multiple pathways and potential synergy among HDAC and EGFR/HER2 inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , ErbB Receptors/antagonists & inhibitors , Histone Deacetylases/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Growth Processes/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Female , HeLa Cells , Hep G2 Cells , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/enzymology , Quinazolines/chemical synthesis , Receptor, ErbB-2/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
Mol Cancer Ther ; 8(12): 3296-306, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952121

ABSTRACT

CUDC-305 is a heat shock protein 90 (HSP90) inhibitor of the novel imidazopyridine class. Here, we report its activities in non-small cell lung cancer (NSCLC) cell lines with gene deregulations conferring primary or secondary resistance to epidermal growth factor receptor (EGFR) inhibitors. We show that CUDC-305 binds strongly to HSP90 extracted from erlotinib-resistant NSCLC cells (IC50 70 nmol/L). This result correlates well with the potent antiproliferative activity in erlotinib-resistant NSCLC cell lines (IC50 120-700 nmol/L) reported previously. Furthermore, it exhibits durable inhibition of multiple oncoproteins and induction of apoptosis in erlotinib-resistant NSCLC cells. CUDC-305 potently inhibits tumor growth in subcutaneous xenograft models of H1975 and A549, which harbor EGFR T790M mutation or K-ras mutations conferring acquired and primary erlotinib resistance, respectively. In addition, CUDC-305 significantly prolongs animal survival in orthotopic lung tumor models of H1975 and A549, which may be partially attributed to its preferential exposure in lung tissue. Furthermore, CUDC-305 is able to extend animal survival in a brain metastatic model of H1975, further confirming its ability to cross the blood-brain barrier. Correlating with its effects in various tumor models, CUDC-305 induces degradation of receptor tyrosine kinases and downstream signaling molecules of the PI3K/AKT and RAF/MEK/ERK pathways simultaneously, with concurrent induction of apoptosis in vivo. In a combination study, CUDC-305 enhanced the antitumor activity of a standard-of-care agent in the H1975 tumor model. These results suggest that CUDC-305 holds promise for the treatment of NSCLC with primary or acquired resistance to EGFR inhibitor therapy.


Subject(s)
Benzodioxoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Benzodioxoles/metabolism , Benzodioxoles/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Erlotinib Hydrochloride , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 15(12): 4046-57, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19509149

ABSTRACT

PURPOSE: We designed and synthesized CUDC-305, an HSP90 inhibitor of the novel imidazopyridine class. Here, we report its unique pharmacologic properties and antitumor activities in a variety of tumor types. EXPERIMENTAL DESIGN: The potency of the compound was analyzed by fluorescence polarization competition binding assay. Its antiproliferative activities were assessed in 40 human cancer cell lines. Its pharmacologic properties and antitumor activities were evaluated in a variety of tumor xenograft models. RESULTS: CUDC-305 shows high affinity for HSP90alpha/beta (IC(50), approximately 100 nmol/L) and HSP90 complex derived from cancer cells (IC(50), 48.8 nmol/L). It displays potent antiproliferative activity against a broad range of cancer cell lines (mean IC(50), 220 nmol/L). CUDC-305 exhibits high oral bioavailability (96.0%) and selective retention in tumor (half-life, 20.4 hours) compared with normal tissues. Furthermore, CUDC-305 can cross blood-brain barrier and reach therapeutic levels in brain tissue. CUDC-305 exhibits dose-dependent antitumor activity in an s.c. xenograft model of U87MG glioblastoma and significantly prolongs animal survival in U87MG orthotopic model. CUDC-305 also displays potent antitumor activity in animal models of erlotinib-resistant non-small cell lung cancer and induces tumor regression in animal models of MDA-MB-468 breast cancer and MV4-11 acute myelogenous leukemia. Correlating with its efficacy in these various tumor models, CUDC-305 robustly inhibits multiple signaling pathways, including PI3K/AKT and RAF/MEK/ERK, and induces apoptosis. In combination studies, CUDC-305 enhances the antitumor activity of standard-of-care agents in breast and colorectal tumor models. CONCLUSION: CUDC-305 is a promising drug candidate for the treatment of a variety of cancers, including brain malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Blood-Brain Barrier/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays
13.
Genes Cells ; 10(6): 503-16, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15938710

ABSTRACT

Embryonic stem (ES) cells have the capacity to differentiate to every cell type that constitutes fetal or adult tissues. To trace and quantitatively assess the differentiation of ES cells into gut endodermal cells, we used an ES cell line with the lacZ gene inserted into the pdx-1 locus. Targeted mutations of pdx-1 in mice demonstrate that pdx-1 is required for pancreatic and rostral duodenal development; therefore, pdx-1 serves as an excellent early gut regional specific marker. When these ES cells were differentiated by removal of leukemia inhibitory factor (LIF), only fractional cells turned into lacZ positive, which indicates pancreatic-duodenal differentiation. Co-cultivation of ES cells with pancreatic rudiments induced a significant increase in the proportion of lacZ positive cell numbers and this increase was further enhanced by forced expression of a chick putative endoderm inducer gene, cmix. Transforming growth factor (TGF)-beta2 mimicked the effects of pancreatic rudiments and this effect was enhanced by cmix expression. Expression analysis showed over-expression of cmix induced endodermal marker genes. These data indicate that one can make use of this knowledge on molecular events of embryonic development to drive ES cells to differentiate into pdx-1 expressing endodermal cells in vitro.


Subject(s)
Endoderm/metabolism , Homeodomain Proteins/metabolism , Signal Transduction , Stem Cells/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Biomarkers , Cell Differentiation/physiology , Cell Line , Chickens , Coculture Techniques , Duodenum/cytology , Duodenum/embryology , Duodenum/metabolism , Embryo, Nonmammalian , Endoderm/cytology , Fibroblast Growth Factors/metabolism , Genetic Engineering , Homeodomain Proteins/genetics , Lac Operon , Mice , Microinjections , Models, Biological , Pancreas/cytology , Pancreas/embryology , Pancreas/metabolism , RNA/metabolism , Stem Cells/cytology , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...